Summary
Liver transplantation is widely indicated as a curative treatment for selected patients with hepatocellular carcinoma. However, with recent therapeutic advances, as well as efforts to increase the donor pool, liver transplantation has been carefully expanded to patients with other primary or secondary malignancies in the liver. Cholangiocarcinoma, colorectal and neuroendocrine liver metastases, and hepatic epithelioid haemangioendothelioma are amongst the most relevant new indications. In this review we discuss the fundamental concepts of this ambitious undertaking, as well as the newest indications for liver transplantation, with a special focus on future perspectives within the recently established concept of transplant oncology.
Keywords: Liver transplant, Oncology, Hepatocellular Carcinoma, Cholangiocarcinoma, Colorectal Liver Metastasis, Neuroendocrine Tumours, Hepatic Epithelioid Hemangioendothelioma
Key points
Liver transplantation is widely indicated as a curative treatment for selected patients with hepatocellular carcinoma.
Increasing the donor pool, liver transplantation has been carefully expanded to patients with other primary or secondary malignancies in the liver.
Cholangiocarcinoma, colorectal and neuroendocrine liver metastases, and hepatic epithelioid haemangioendothelioma are amongst the most relevant new indications.
Alt-text: Unlabelled Box
Introduction
Annually, hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) are diagnosed in 841,000 people and are responsible for 782,000 deaths worldwide.1 Colorectal cancer is diagnosed in 1.8 million people every year and it is estimated that ~50% of these patients will develop colorectal liver metastasis (CRLM).[1], [2] For patients with liver cancer, the surgical removal of the tumour offers the best chance of cure. Unfortunately, only a minor proportion of these patients are candidates for liver resection (LR) mostly because of decompensated liver disease. Liver transplantation (LT) offers a chance of cure given that it removes the tumour with the widest margin, as well as removing the pro-carcinogenic hepatic microenvironment.
Transplantation as treatment for unresectable liver cancer has been explored since the early development of LT.3 The initial experiences with LT for liver cancer were, however, disappointing.[4], [5], [6] The landscape of LT for cancer changed in 1996, when a strict selection criteria for patients was published.7 Since then, with better patient selection and refinements to operative and postoperative care, LT has become an effective treatment for several hepatic malignancies. Together with other important advances in hepatology and oncology (e.g. new chemotherapies for gastrointestinal cancer; direct-acting antivirals [DAA] for hepatitis C) a new field in medicine has risen: transplant oncology.8 In this review, we aim to explore the current indications for LT as a treatment for hepatic malignancies, with a special focus on future perspectives within the concept of transplant oncology.
Hepatocellular carcinoma
The treatment of HCC has become multidisciplinary, involving hepatobiliary and transplantation surgery, hepatology, interventional radiology, radiation and medical oncology. Among all possible strategies to treat HCC, LT offers the best chance of cure.9 Unfortunately, the number of available grafts is insufficient for all potential candidates. For this reason, LT is reserved for patients who will benefit most. Efforts should focus on strategies to better select patients and to increase the number of available grafts.
LT for HCC: improvement of patient selection
Selection criteria worldwide
Patient selection is the mainstay of LT for cancer. After the Milan criteria were published, LT became the standard of care for patients with unresectable HCC who fit within its bounds.7 The success of the Milan criteria has led to increased interest in expanding the criteria for LT.10 Several “expanded criteria” have been proposed over the last 10 years (Table 1).11
Table 1.
Liver transplantation criteria for patients with hepatocellular carcinoma.
| Criteria | Definition | Recurrence-free survival | Post-transplantation survival | Innovation |
|---|---|---|---|---|
| Milan criteria (MC)7 | Single tumour ≤5 cm or 3 tumours all ≤3 cm | 92% at 4 years | 85% at 4 years | First criteria widely accepted |
| UCSF criteria153 | Single tumour ≤6.5 cm or 3 tumours all ≤4.5 cm with TTD ≤8 cm | 90.9% at 5 years | 80.9% at 5 years | Extended MC criteria limits |
| Up-to-7 criteria154 | The sum of the maximum tumour diameter and number ≪7 | Beyond MC but within Up-to-7: 64.1% at 5 years | Beyond MC but within Up-to-7: 71.2% at 5 years | Extended MC limits |
| TTV13 | TTV ≤115 cm3 Serum AFP ≤400 ng/ml |
Beyond MC but within TTV/AFP: 68% at 4 years | Beyond MC but within TTV/AFP: 74.6% at 4 years | Added surrogates for tumoural biology |
| Extended Toronto criteria (ETC)20 | No limit in size and number No vascular invasion No extrahepatic disease No cancer-related symptoms Biopsy of largest tumour not poorly differentiated |
Cumulative risk of recurrence for patients beyond MC but within ETC: 30% at 5 years | Beyond MC but within ETC: 68% at 5 years | Added surrogates for tumoural biology Extended MC limits. |
| Kyoto criteria21 | Number of lesions ≤10 tumours Size biggest lesion ≤5 cm DCP ≤400 mAU/ml |
Cumulative risk of recurrence of patients beyond MC but within Kyoto: 30% at 5 years | Beyond MC but within Kyoto: 65% at 5 years | Added serum biomarker to the criteria (DCP) |
| 5-5-50022 | Size biggest tumour size ≤5 cm Number of lesions ≤5 Serum AFP ≤500 ng/ml |
71.4% at 5 years | 74.8% at 5 years | Identified patients with worse prognosis within MC. |
AFP, alpha-fetoprotein; DCP, des-gamma-carboxyprothrombin; TTD, total tumour diameter; TTV, total tumour volume; UCSF, University of California at San Francisco.
Impact of serum alpha-fetoprotein
Serum alpha-fetoprotein (AFP) is an important biomarker in HCC. The 5-year disease-free survival (DFS) for patients with a serum AFP ≫1,000 ng/ml has been reported as 53% in comparison with 80% in patients with AFP ≤1,000 ng/ml.12 Toso et al. demonstrated that patients beyond Milan criteria and with AFP ≤400 ng/ml can achieve satisfactory post-LT outcomes.13 The hazard ratio (HR) of HCC recurrence for patients with total tumour volume (TTV) ≤115 cm3 and serum AFP ≪400 ng/ml was 2.0 (95% CI 1.7–2.4), when compared to patients with TTV ≫115 cm3 and serum AFP ≫400 ng/ml.[14], [15] The “AFP model” uses a scoring system to classify patients by their risk of recurrence based on largest tumour diameter, number of lesions and serum AFP. Patients who have ≤2 points have a lower probability of recurrence and are within the criteria. Among these patients, the 5-year recurrence rate was 14% vs. 48% for those beyond the AFP criteria.15 The Metroticket 2.0 system applies serum AFP, tumour size and tumour number to determine the risk of HCC-related death after LT (applying competing-risk analysis). The c-statistic of the model was 0.72, which was superior to previous criteria.16 Halazun et al. recently published a model incorporating the concept of AFP response during waiting time. The AFP response was defined as the difference between the highest value and the final pre-LT serum AFP. They showed that dynamic changes in AFP during waiting time are valuable tools to identify patients beyond Milan criteria who could have good outcomes after LT.17
Surrogates of tumour biology
Surrogates of tumour biology have been studied with the aim of improving the selection criteria for HCC. Tumoural differentiation has been proposed as a selection criteria for LT.[18], [19] Sapisochin et al. prospectively demonstrated that in the absence of macroscopic vascular invasion, extrahepatic disease, cancer-related symptoms and poor differentiation (the Extended Toronto Criteria) patients can undergo LT with satisfactory results regardless of tumour size and number (Table 1).20 Kaido et al. have shown the utility of associating the levels of serum des-γ-carboxy prothrombin (DCP) to size and number of tumours.21 The Kyoto criteria select patients with a DCP ≤400 mAU/ml, a largest tumour diameter ≤5 cm and ≤10 lesions (Table 1). Recently, the 5-5-500 criteria (tumour size ≤5 cm, tumour number ≤5, and AFP ≤500 ng/ml) was associated with a 5-year recurrence rate of 7.3% in patients treated with LDLT.22 The use of 18F-fluorodeoxyglucose positron-emission tomography/CT (18FDG-PET/CT) has been correlated with HCC recurrence and increasingly used as a tool for patient selection.[23], [24] Further research is still needed to be able to incorporate PET/CT widely into clinical practice.
Time on the waitlist has been considered a surrogate of tumour biology.25 Recently, the United Network for Organ Sharing (UNOS) implemented a 6-month mandatory observation period prior to granting MELD exception points.26 However, given the high heterogeneity in referral times amongst centres worldwide it is difficult to extrapolate a threshold to all jurisdictions. Firl et al. validated the hazard associated with LT in HCC (HALT-HCC) and demonstrated a significant heterogeneity by site and year, reflecting practice trends over the last decade..27
Response to locoregional therapies
Response to locoregional therapies (LRT) such as ablation, transarterial chemoembolisation (TACE), selective internal radiation therapy and stereotactic body radiation therapy correlates with tumour biology.[28], [29], [30] Complete pathological response in the explant has been associated with a higher overall survival (OS) and DFS.30 In patients within the Milan criteria, poor response to LRT was associated with HCC-dependent transplant failure. Lai et al. have shown that patients with progressive disease despite LRT had a higher risk of dropout or post-transplant HCC recurrence (subdistribution HR 5.62, 95% CI 4.10–7.69).31 Additionally, Mehta et al. demonstrated significantly improved post-LT outcomes when restricting LT to patients with a reduction in AFP from ≫1,000 to ≪500 ng/ml after LRT.32 In the near future, the assessment of tumour response on pre-LT imaging can be improved with artificial intelligence methods (e.g. radiomics).[33], [34], [35]
Primary vs. salvage LT
The optimal approach for patients who have failed on prior curative treatments for HCC is controversial. In retrospective series, salvage LT and secondary LR had similar outcomes.[36], [37] However, the risk of recurrence after salvage LT may be lower than secondary resection.[38], [39] The decision to treat with secondary resection or salvage LT remains controversial and depends on the availability of organs within each jurisdiction.
Genetic advances in HCC
Profiling the genomic and biological patterns of tumours and correlating them with clinical outcomes is key to better understanding HCC biology.40 There are a wide range of HCC biomarkers currently under investigation, mostly in phases I and II studies. MacParland et al. using single-cell RNA techniques have shown that there are at least 2 different types of immune cells in the liver, and this may be key for HCC-directed therapies.41 Different authors have published a wide range of possible biomarkers in HCC diagnosis and surveillance, such as osteopontin,42 GALAD score and BALAD-2,43 midkine,44 DCP, lectin-bound alpha-fetoprotein (or AFP-L3),45 Dickkopf-1, glypcan-3, HCCR, alpha-L-fucosidase,[46], [47] golgi protein-73, squamous-cell carcinoma antigen (or SCC-IgM),[48], [49] micro-RNA, kininogen,[50], [51] metabolomics, proteomics,[52], [53] circulating tumour cells and cell-free DNA,54 polo-like kinase genes, PD-1 (programmed cell death protein 1) and TIM-3 (T-cell immunoglobulin and mucin-domain containing-3).[55], [56] Most of this research has not been done in the transplant population. Table 2 summarises the main biomarkers under research.
Table 2.
Biomarkers under research for HCC diagnosis and treatment beyond liver transplantation that could be utilised in the transplant setting.
| Authors | Year | Biomarker | Phase | Applicability |
|---|---|---|---|---|
| Mehta et al.40 | 2018 | AFP | V | Diagnosis and survival |
| Marrero et al.45 | 2009 | DCP | III | Diagnosis and survival |
| Fedarko et al.42 | 2001 | Osteopontin | III | Diagnosis |
| Vongsuvanh et al.44 | 2016 | Midkine | III | Diagnosis |
| Berhane et al.43 | 2016 | GALAD score | III | Survival |
| Marrero et al.45 | 2009 | AFP-L3 | II | Diagnosis and survival |
| Jang et al.155 | 2016 | Dickkopf-1 | II | Diagnosis |
| Qiao et al.46 | 2011 | Glypican-3 | II | Diagnosis |
| Giardina et al.47 | 1992 | Alpha-1-fucosidase | II | Diagnosis |
| Ismail et al.48 | 2017 | Golgi protein-73 | II | Diagnosis and surveillance |
| Pozzan et al.49 | 2014 | Squamous cell carcinoma antigen | II | Diagnosis and surveillance |
| Shi et al.50 | 2015 | Micro RNA | I-II | Diagnosis |
| Wang et al.52 | 2013 | Metabolomics | I | Diagnosis |
| Sengupta et al.53 | 2013 | Proteomics | I-II | Diagnosis |
| Pantel et al.54 | 2017 | Cell-free DNA | I | Diagnosis |
| MacParland et al.41 | 2018 | Single-cell RNA | I | Treatment |
| Pellegrino et al.55 | 2010 | Polo-like kinases | I | Diagnosis |
| Sangro et al.156 | 2013 | CTLA-4 | I-III | Treatment |
| Li et al.56 | 2016 | PD-1 and TIM-3 | I | Diagnosis |
| Sun et al.157 | 2018 | PDL-1 | I | Treatment |
AFP, alpha-fetoprotein; AFP-L3, alpha-fetoprotein-lecithin 3; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; DCP, des-gamma-carboxy prothrombin; GALAD, acronym for: Gender, Age, AFP-L3, AFP, DCP; PD-1, programmed cell death protein 1; PDL-1, programmed death-ligand 1; TIM-3, T-cell immunoglobulin and mucin-domain containing-3.
Genomic expression does not always reflect an immunologically active phenotype in patients with HCC. Thus, a tumour biopsy may not provide all the information necessary for therapeutic decision making. The association of these tumour genetic findings with adjacent normal liver assessment, serum circulating tumour DNA (ctDNA) and the phenotypic expression in metabolites or serum proteins, could potentially identify a more aggressive tumour behaviour, changing the therapeutic indication and selection for transplantation. At this point, this is all hypothetical and further research is needed and ongoing in this area.
LT for HCC: Increasing the donor pool
To support the expansion of LT for patients with HCC without compromising patients without HCC, there is a need to increase the number of available grafts. There is controversial data on the impact that the different types of grafts can have on the outcomes of patients transplanted with HCC.
Use of marginal grafts
The use of marginal grafts (i.e. older donors, donors after cardiac death (DCD), split livers, steatotic grafts or hepatitis C virus (HCV)-infected grafts) is one of the options to increase the donor pool. Marginal grafts have been of particular interest for patients with HCC given that they usually have better liver function than patients listed with decompensated cirrhosis. The use of non-ideal grafts (i.e. “liver that nobody wants”) in patients with HCC has been investigated, showing that acceptable outcomes can be achieved.57 However, this strategy must be approached with caution to avoid putting patients in good general condition at higher risk of post-transplant complications. Initial studies with the use of DCD grafts raised questions about the increased risk of tumoural recurrence due to the potential oncogenic effect of ischaemia-reperfusion injury.[58], [59] This concern was not confirmed by subsequent studies.[60], [61], [62] Likewise, the use of grafts from older donors was seen as a risk factor for post-LT HCC recurrence63 and currently donor age is not by itself a limitation for donation in many centres worldwide.[64], [65] The use of HCV-infected grafts in recipients with HCV has been proven safe.[66], [67] Cotter et al. demonstrated that, in the DAA era, there has been an increase in the utilisation of HCV-viraemic donor livers, including into HCV-negative recipients, with good graft outcomes.68 The use of DAAs has changed the landscape of HCV treatment and, annually, less patients with end-stage liver disease due to HCV are listed for LT.[68], [69], [70], [71], [72], [73], [74]
Living donor liver transplantation
The most important intervention to increase the donor pool is living donor liver transplantation (LDLT). Initial results on LDLT for HCC indicated an increased risk of recurrence.[75], [76] However, more recent studies did not confirm this finding. In an intention-to-treat analysis, 2 studies have shown similar outcomes between patients who underwent LDLT and those undergoing LT with grafts from brain-death donors (DDLT).[77], [78] Goldaracena et al. have shown that LDLT is associated with survival benefit for patients with HCC. In an intention-to-treat analysis, patients who had a potential living donor had a 5-year OS rate of 68% compared to 57% in patients without a potential donor.79 The presence of a potential live donor was a protective factor for death (HR 0.67; 95% CI 0.53–0.86). Despite being an excellent strategy for patients with HCC waiting for a LT, the widespread use of LDLT must be limited to centres that perform high volumes of both advanced hepatobiliary surgery and LT to diminish the risk of complications for the donor.80 After live donation, the rate of overall postoperative complications is reported to be around 25–30%, with major complications occurring in 9–10% of patients.[80], [81] Donor mortality has also been reported and estimated between 0.1–0.3%.[82], [83]
LT for HCC: future prospects
The future direction of LT for HCC will focus on the identification of patients at higher risk of recurrence to prevent futile transplantation. This selection will likely move away from tumour size and number, and allow for the incorporation of surrogates of tumour biology. The use of imaging methods such as 18FDG-PET/CT or genomic technics that could identify circulating DNA or single-cell RNA as a genetic signature of recurrence may improve our current criteria for patient selection. Radiomics applied to pre-treatment imaging assessments may enable clinicians to predict tumour behaviour in the near future. Xu et al. have demonstrated its ability to identify the presence of microvascular invasion in 495 patients with resected HCC.35
In the context of LT, neoadjuvant therapies may also increase access to transplantation for patients who are currently not candidates. An example of this approach would be patients with macrovascular invasion who respond to neoadjuvant therapies and have a stable period of observation.84 This should only be done under investigational protocols at this time.
Cholangiocarcinoma
Cholangiocarcinoma represented 2% of all LTs performed for malignancies in Europe between 1988–2016.85 Hilar CCA (hCCA) and intrahepatic CCA (iCCA) have distinct molecular pathogenesis and biological behaviour and therefore are presented here as separated entities.
LT for CCA: initial experience
The initial experience with LT for CCA was disappointing. In 1988, the group from Kings College published a series of 93 patients who underwent LT for several malignancies of whom 26 had CCA (13 hilar and 13 intrahepatic).86 The 5-year OS rate for this cohort was 10%.86 In 1997, Pichlmayr et al. published a series of 24 patients with iCCA and 28 with hCCA who underwent LT.87 The 5-year OS rates were 0% and 18% for patients with iCCA and hCCA, respectively.87 This poor initial experience was explained mainly by the lack of criteria for patient selection and the absence of standardised pre- and postoperative treatment.
LT for hCCA
The first study with a strict patient selection and a neoadjuvant therapy protocol was published in 2000 by De Vreede et al..88 The so-called Mayo protocol consists of neoadjuvant treatment with 5-fluoracil (for radiosensitisation) and oral capecitabine (maintenance therapy) until LT is performed with preoperative external beam radiation therapy and local brachytherapy.89 Gemcitabine along with capecitabine are applied in the neoadjuvant protocol in other centres.90 The effectiveness of LT for hCCA was validated in North America by a study reporting on data from 12 US centres, including 287 patients who underwent LT, in which a 5-year DFS rate of 65% was achieved.91 Mantel et al. have assessed the results of LT for hCCA in a cohort from the European Liver and Intestine Transplant Association (ELITA) and showed a 5-year OS rate of 59%.92 After these satisfactory results, hCCA became an indication in many jurisdictions worldwide.[93], [94], [95], [96], [97]
The use of LT for patients with locally advanced hCCA and a low preoperative probability of achieving complete resection (e.g. tumours ≫3 cm and/or with ipsilateral intrahepatic portal branch invasion and/or positive lymph nodes) has been the topic of debate. One small retrospective study (13 patients in the LT group and 7 patients in the resection group) has shown superior results for LT.98 The group from Nagoya published a series of 216 patients with type IV hCCA who were treated by resection.99 The 5-year OS rate was 53% among those patients without lymph nodal metastasis.99 The authors argue that this OS rate is comparable to that seen after LT for hCCA; but, unfortunately, this study did not have an LT group for comparison. Ethun et al. retrospectively compared the outcomes of LR and LT in 304 patients. Resection was attempted in 234 patients and successful in 191 (82%). In the LT group, 70 patients were listed and 46 (66%) underwent LT. Transplantation was associated with improved OS compared to LR (64% vs. 18% 5-year OS, p ≪0.001).100 These results need to be analysed with caution given the observational nature of the design. Aiming to provide a definitive answer on the effectiveness of LT for patients with locally advanced hCCA, the TRANSPHIL trial (NCT02232932) is currently recruiting patients. This is a prospective, randomised, multicentre study comparing neoadjuvant chemoradiation plus LT to LR in patients with resectable hCCA.101 Results are expected in 2021.
LT for iCCA
LR is the first treatment option for iCCA. Since the early 2000s, several publications have shown that LT might be an option for patients with iCCA who are not candidates for LR. Robles et al. assessed 23 patients who underwent LT for HCC and were diagnosed with iCCA in the explant. The cohort had 13 (57%) patients with early/intermediate stage iCCA and 10 (43%) with advanced disease.102 The median OS was significantly higher for patients with early/intermediate stage (60 months) compared to patients with advanced disease (22 months), p = 0.048.102 Sapisochin et al. assessed a similar cohort of patients who underwent LT.103 Among 29 patients, the 5-year OS rate was 45%.103 However, patients with very early iCCA (defined as tumours ≤2 cm) had significantly lower 5-year risk of recurrence (18% vs. 65%, p = 0.01) and greater 5-year OS (65% vs. 45%, p = 0.02) than those with multifocal and larger tumours. Vilchez et al. studied 4,049 patients who underwent LT for malignancies from the UNOS database.104 Of these patients, 3,515 patients had HCC, 440 had iCCA and 94 had mixed HCC-CCA in the explant. The 5-year OS rate was 62% for patients with HCC, 47% for patients with iCCA and 40% for patients with mixed HCC-CCA (p = 0.02).104 Unfortunately, this study did not address the outcomes according to the tumour burden. The benefit of LT for patients with early stages of iCCA was confirmed in an international collaborative study. Among 48 patients with iCCA, the 5-year cumulative risk of recurrence was 18% for those with very early iCCA and 61% for those with more advanced disease (p = 0.01).105 The 5-year OS rate between the very early iCCA and advanced iCCA was 65% and 45%, respectively (p = 0.02).105 These retrospective studies demonstrated that patients with very early iCCA, who are not candidates for LR, might have acceptable OS with LT. The application of LT for patients with unresectable very early iCCA still requires validation by a prospective study. The validation study is currently recruiting (NCT02878473) and results are expected within 5 years.106 Until further investigation, iCCA should remain a contraindication for LT outside of clinical trials.
In the non-cirrhotic population, Lunsford et al. have published a prospective case series of 21 patients with iCCA who were assessed for LT.107 This series had a well-defined neoadjuvant protocol. Inclusion criteria were solitary tumour greater than 2 cm or multifocal disease confined to the liver without radiological evidence of macrovascular or lymph nodal involvement. Among the initial 21 patients, 12 were listed for LT and 6 underwent LT. The 6 recipients were followed for a median of 36 months and 3 had CCA recurrence.107 This approach of neoadjuvant chemotherapy with or without radiation therapy could be useful for downstaging therapy in patients with unresectable CCA or as a selection criteria for LT.[91], [107] Le Roy et al. have studied patients with unresectable iCCA who received neoadjuvant chemotherapy. Of 74 unresectable patients, 39 (53%) patients were successfully downstaged and underwent LR.108 The use of radioembolisation with yttrium-90 (Y90) has been investigated as an option for downstaging and/or neoadjuvant therapy before LT. In a study from Rayar et al., patients with unresectable iCCA were treated with Y90 combined with systemic chemotherapy. In this study, 8/45 patients were successfully converted to resection.109 The use of neoadjuvant therapies to convert unresectable patients might be preferable to LT in light of organ scarcity. However, even though some patients could be successfully downstaged for resection, it would be fair to offer LT to patients who remained unresectable in the absence of disease progression during neoadjuvant treatment. Therefore, future studies assessing neoadjuvant therapies for advanced iCCA should aim both to downstage patients for resection and to select patients for LT.
LT for CCA: future perspectives
A better assessment of patients that have aggressive tumoural biology or extrahepatic disease is key to avoid futile transplantation. For example, the presence of positive circulating tumoural DNA seems to be related to prognosis.110 Genetic sequencing is also a very important tool in selecting patients with a lower likelihood of recurrence. Mutations in KRAS, BAP1 and CDKN2A are related to a higher probability of recurrence, while mutations in FGFR2 are related to more indolent phenotype.[111], [112], [113] In hCCA, mutations in P53, BRCA1, BRCA2 and PIK3CA are related to a worse prognosis.[111], [112] Whether these genetic profiles will be applied as a selection tool in LT for CCA is still under investigation and cannot be recommended.
The future of CCA treatment lies in the development of specific drugs directly targeting pathways of carcinogenesis. Several biomarkers are being studied, opening up opportunities for translational research initiatives in CCA. It is increasingly evident that the CCA desmoplastic microenvironment plays an important role in cancer cell development, and strategies targeting the tumour stroma in combination with the CCA cancer cell will present new diagnostic and therapeutic perspectives.114
Due to the rarity of this tumour, initiatives are necessary to develop international consortia such as the Thailand Initiative in Genomics and Expression Research for Liver Cancer (TIGER-LC), the European Network for the Study of Cholangiocarcinoma (ENS-CCA), the International Cholangiocarcinoma Research Network, along with patient advocacy groups like the Cholangiocarcinoma Foundation, to enable the creation of international translational and clinical research collaborations that can perform multicentre clinical trials with the aim of elucidating new therapies.
Colorectal liver metastasis
LR is the only curative treatment for CRLM. Recent advances in medical and surgical treatments have allowed for an important expansion in the limits of resectability and life expectancy in this population.115 Only 30–40% of patients are candidates for LR at the time of disease presentation.116 The main reason for precluding LR in patients with CRLM is insufficient liver remnant volume. For patients with insufficient liver remnant and no extrahepatic involvement, LT is becoming an option given that total hepatectomy will remove all viable disease.
LT for unresectable CRLM: A new hope
Initial reports on the use of LT for unresectable CRLM showed poor results. In 1991, Mühlbacher et al. reported their experience with 17 patients transplanted for CLRM, showing a 5-year OS rate of 12% and a 60% recurrence rate.117 To improve outcomes they restricted LT to patients with negative lymph node disease in the primary specimen.118 Penn published the results from a North American cohort.5 This was a retrospective report of 637 patients with liver cancer; of those 8 patients underwent LT for CRLM. The recurrence rate was 70% and the 30-day mortality was 11%. Due to these poor results, in the early 1990s the use of LT for CLRM was abandoned.
The use of LT for CRLM has regained momentum after the work of Hagness et al..119 Scandinavia is a region where the liver graft offer exceeds the demand.120 In the SECA-I (SEcondary CAncer I) study, 21 patients underwent LT for CRLM.119 The OS rate was 95% at 1 year and 60% at 5 years; the DFS rate was 35% at 1 year. Nineteen of 21 patients had tumour recurrence after a median 6 months (range 2–24 months). The most common site of recurrence was pulmonary (17/19 patients). In a subsequent publication, the authors assessed the recurrence patterns, showing a 57% 5-year post-recurrence survival. Patients with pulmonary-only metastasis, had slow growing recurrences despite immunosuppression, allowing for resection in 9/13 patients.121 The remaining 8/17 recipients developed metastases in multiple sites, including hepatic recurrence, which was associated with the worst outcomes.121 In the SECA-I study, the exclusion criteria were not very restrictive. The exclusion criteria were presence of extrahepatic disease and weight loss ≫10%. This approach allowed for the isolation of independent factors predicting worse OS: carcinoembryonic antigen (CEA) ≫80 ug/L, progression of the metastases under neoadjuvant chemotherapy, tumour diameter ≫5.5 cm, time interval from resection of the primary to LT ≪2 years.119 An international consortium published the results of 12 patients with CRLM who underwent LT.122 The OS rate was 50% with 6 patients having cancer recurrence after a median follow-up of 26 months. In accordance with previous studies, the most common site of recurrence was pulmonary.
LT for CRLM: beyond the initial enthusiasm
As the concept of transplant benefit is gaining recognition over classic survival after transplantation or simplistic urgency criteria,[123], [124] LT for CLRM will likely find its place in future practice. However, before it becomes a recognised indication, definitive evidence is required to address a few outstanding issues:
It has to be proven that transplantation is superior to chemotherapy
The SECA-I study provided encouraging evidence in favour of transplantation. Aiming to compare the results after LT to those seen after palliative chemotherapy, Dueland et al. compared the outcomes of their transplanted population (21 patients) to a matched cohort of patients who underwent palliative therapy.125 They demonstrated improved 5-year OS in favour of LT (56%) compared to the chemotherapy (9%).125 The cost-effectiveness of LT for CRLM in highly selected patients was recently shown.126
Definitive confirmation of these retrospective findings will hopefully come from several ongoing trials. The SECA-III trial (NCT03494946) will compare LT to best multimodal alternative treatment (chemotherapy +/- locoregional therapies). The TRASNMET trial (NCT02597348) is a multicentric trial comparing LT for unresectable CRLM to chemotherapy only. Our centre is currently enrolling patients in a pilot study to assess the safety and effectiveness of neoadjuvant chemotherapy followed by LDLT for patients with unresectable CRLM (NCT02864485).
Patient selection has to be refined
The population of patients enrolled into the SECA-I study was quite heterogeneous, helping with the identification of 4 factors associated with better survival (see above). Low CEA levels were also confirmed as a good prognostic factor by another study from an international consortium.122 Moreover, a retrospective analysis using the SECA-I data was able to select a low-risk population (Oslo score 0-3) with a 5-year OS rate of 75%.127 Another retrospective analysis on the SECA study data helped identify other predictors of post-transplant OS, such as the ‘metabolic tumour volume’ and ‘the total lesional glycolysis’ of the CLRM measured by 18F-FDG PET/CT,128 which could have a role in identifying patients with minor extrahepatic disease.129 The recently published SECA-II trial showed that response to neoadjuvant chemotherapy is, in fact, important. Patients with a minimal response to chemotherapy of 10% had OS rates of 100%, 83% and 83% at 1-, 3- and 5-years , respectively.130 The TRANSMET study contemplates additional criteria such as BRAF mutations, in order to exclude patients with aggressive tumour biology. This is also an exclusion criterion in the Toronto trial.
A standardised chemotherapy protocol has to be defined
Thanks to modern neoadjuvant chemotherapy protocols, around 10–15% of patients with initially unresectable CRLM become candidates for LR.[131], [132] Therefore, it is clear that upfront chemotherapy should be offered to every patient potentially considered for LT, with the aim of conversion. In addition, as supported by the SECA-I and SECA-II trials, poor response to chemotherapy might be a criterion to identify high-risk candidates, who may not benefit from LT. Whether or not it is beneficial to administer post-transplant chemotherapy, instead, is a point yet to be explored. Patients enrolled in the SECA-I study were not given adjuvant (post-transplant) chemotherapy. The SECA-II/III and RAPID (see Table 3) trials do not have it as a formal requirement. With the exception of the SECA-II study, all the patients enrolled in these trials undergo liver transplant after multiple cycles of chemotherapy, some of them having already received second- and third-line treatments. In this context, the benefit of additional cycles may be marginal compared to their toxic effects, especially when involving small grafts (RAPID, LIVERT(W)OHEAL) and liver regeneration should not be impaired. Patients enrolled in the TRASMET study receive limited post-transplant chemotherapy, while in the trial from Toronto, adjuvant standard-of-care chemotherapy is given. This last study will provide the more valuable information about the real benefit of post-transplant chemotherapy in the context of LT for CRLM, although only a randomisation would provide definitive evidence.
Table 3.
Ongoing studies on LT for CRLM.
| Study Name Sponsor NCT |
Study design | Start/end estimated year | Patients enrolled(n) | Arms | Outcome(s) | Main inclusion criteria | Main exclusion criteria |
|---|---|---|---|---|---|---|---|
| SECA II Oslo University Hospital NCT 01479608 |
Clinical trial monocentric randomised (A) Open label |
2012 / 2025 | 25 |
A1: Transplantation vs. A2: resection (randomised) B: Liver transplantation For non-resectable patients metachronous disease C: Liver transplantation For non-resectable patients synchronous disease |
OS (10 years) | Histologically verified adenocarcinoma in colon/rectum No signs of extrahepatic metastatic disease/local recurrence (PET/CT) Received at least 3 cycles of chemotherapy with no increase in size of the lesions (A only) Six or more liver metastases technically resectable |
Weight loss ≫10% the last 6 months Patient BMI ≫30 |
| TRANSMET Paris Hospitals NCT 01479608 |
Clinical trial multicentric randomised open label |
2015 / 2027 | 90 | Intervention: liver transplantation vs. Non-intervention: non-experimental standard chemotherapy |
OS (5 years) DFS Quality of life |
Histologically verified adenocarcinoma in colon/rectum Liver metastases, not amenable to liver resection ≥ 3 months of tumour control during the last chemotherapy line BRAF wild-type CRC on primary tumour or liver metastases ≤ 2 lines of chemotherapy for metastatic disease. |
General contraindication to LT Patients not having received standard treatment for the primary CRC according to recommended guidelines |
| SECA III Oslo University Hospital NCT 03494946 |
Clinical trial monocentric randomised open label |
2016 / 2027 | 30 | Intervention: liver transplantation vs. Comparator: any other treatment (including chemotherapy, ablation, TACE, SIRT) |
OS (2 years after randomisation) | Histologically verified adenocarcinoma in colon/rectum Liver metastases, not amenable to liver resection All patients should have progressive disease according to RECIST criteria, or intolerance to 1st line chemotherapy No signs of extrahepatic metastatic disease, except patients may have 1-3 resectable lung lesions all ≪15 mm |
Weight loss ≫10% the last 6 months Patient BMI ≫30 Liver lesion≫10 cm Three negative prognostic factors at time of randomisation (CEA≫80, less than 2 years from diagnosis, diameter of largest liver lesion ≫5.5 cm) |
| RAPID Oslo University Hospital NCT 02215889 |
Clinical trial single group assignment |
2014 / 228 | 20 | Intervention: 2-stage total hepatectomy + liver transplantation of segments 2/3 from deceased donor | Percent of transplanted patients receiving second stage hepatectomy within 4 weeks of segment 2/3 transplantation. OS (5 years) |
Histologically verified adenocarcinoma in colon/rectum Liver metastases, not amenable to liver resection Received at least 3 cycles of chemotherapy No signs of extrahepatic metastatic disease, except patients may have 1-3 resectable lung lesions all ≪15 mm |
Weight loss ≫10% the last 6 months Patient BMI ≫30 |
| LIVERT(W)OHEAL Jena University Hospital NCT 03488953 |
Clinical trial single group assignment |
2018 / 2023 | 40 | Intervention: 2-stage total hepatectomy + liver transplantation of segments 2/3 from living donor | OS 3 years after 2nd-stage of hepatectomy (3 years) DFS 3 years after 2nd-stage of hepatectomy (3 years) |
Non-resectable colorectal liver metastases without extrahepatic tumour burden, except resectable pulmonary metastases Stable disease or regression after at least 8 weeks of systemic chemotherapy |
General contraindication to LT |
| University Health Network, Toronto NCT 02864485 | Clinical trial single group assignment |
2016 / 2023 | 20 | OS (5 years) DFS (5 years) |
Bilateral and non-resectable CRLM Received at least 3 cycles of chemotherapy with proven stable disease Primary CRC tumour stage is ≤T4a |
General contraindication to LT BRAF+ tumours |
BMI, body mass index; CEA, carcinoembryonic antigen; CRC, colorectal cancer; CRLM, colorectal liver metastasis; DFS, disease-free-survival; LT, liver transplant; OS, overall survival; PET/CT, positron-emission tomography/CT; SIRT, selective internal radiation therapy; TACE, transarterial chemoembolisation.
Current ongoing trials in the field of LT for CRLM are summarised in Table 3.
LT for CRLM: future perspectives
Coping with a potentially very high demand
LT is a victim of its own success, with already accepted indications exhausting a very limited resource. If the ongoing trials confirmed a superior benefit of LT for unresectable CRLM over other treatments, organ allocation policy will have to deal with a considerable problem. Fortunately, most of the centres where LT for CRLM will become an option, are testing different strategies to mitigate this issue.
Some centres have proposed the use of auxiliary grafts in 2-staged procedures. The so-called RAPID procedure (Resection And Partial Liver Segment 2/3 Transplantation With Delayed Total Hepatectomy) was designed by the Oslo group (NCT02215889).133 It aims to perform a left lateral hepatectomy with a left lateral segment graft implantation. The rationale is to delay the completion of the total hepatectomy to allow the graft to grow. The first step is a limited segment 2-3 resection, which leaves the room for the auxiliary graft from a deceased donor. After reperfusion, the right portal vein is clamped and subsequently ligated if the pressure does not exceed 20 mmHg (if not, other measures are undertaken to lower the pressure: portal banding instead of complete interruption, splenic artery ligation, porto-caval shunting). The graft’s volume increase is assessed regularly until liver/body weight ratio reaches 0.8. At that point patients undergo a second procedure, with totalisation of the hepatectomy. The LIVERT(W)OHEAL study (NCT03488953) from 2 German university hospitals, applies the RAPID concept to live donation.
Transplantation of patients with resectable CRLM
R0 surgical resection is the gold standard treatment for patients with resectable CRLM. Recently, thanks to the advent of extremely effective chemotherapy protocols, even R1 resections can be considered curative if patients have positive response to systemic treatments.[134], [135] Interestingly, very large series on LR for CRLM showed 5-year OS rates of ≪40% for patients presenting with more than 3 metastases,136 which is inferior to the overall 60% OS rate at 5 years of patients enrolled in the SECA-I study (median 8 metastases). On the other hand, the SECA-I population had very stable disease on chemotherapy, in contrast with the large case series on LR that included a broad heterogeneity of cases.
The feeling is that some selected patients with borderline resectable disease and large tumour burden may benefit more from transplantation than from resection.
Neuroendocrine tumours
Liver metastases are common in neuroendocrine tumours (NETs) arising from the small intestine and pancreas.137 Patients with unresectable NET metastases to the liver are candidates for LT if their tumours have low biological aggressiveness. NET represents 0.3% of the LTs performed in Europe, according to the European Liver Transplant Registry.138 The level of evidence on the use of LT for NET metastasis is not high given the absence of information about DFS. In many studies there is a lack of uniform follow-up and assessment of quality of life.139 Therefore, there is certain controversy on the selection criteria for LT and the best time to perform LT in patients with unresectable NET liver metastases. In 2007, Mazzaferro et al. published the most widely used criteria for the selection of patients with NETs for LT: low-grade tumour (G1-G2 or Ki-67 less than 5–10%), primary tumour drained by the portal system completely removed, metastatic diffusion to less than 50% of liver volume, stable disease for at least 6 months with medical therapies and age lower than 60 (relative criteria).140 These criteria are very similar to those adopted by UNOS.141 In Europe, LT for patients with NETs was assessed by Le Treut et al. in 2013.142 Among 213 patients from the European Liver Transplantation Association Registry, they identified a 5-year OS rate of 73%.142 Risk factors for worse outcomes were primary tumour arising from the pancreas, resection of the primary tumour during the LT, presence of hepatomegaly, hepatic involvement ≫50%, tumour bulk, poor differentiation, margin-positive and presence of lymph node involvement.142 Worse survival in patients with pancreatic NETs was also reported by van Vilsteren et al..143 In 2016, the group behind the Milan criteria published a retrospective study showing the long-term results of applying these criteria. They compared 42 patients who underwent LT to 46 who received other therapies in a retrospective cohort. The 5-and 10-year OS rates were 97% and 89%in the LT group and 51% and 22.4% in the control group, respectively (p ≪0.001). The HR for death was 7.4 (95% CI 2.4–23.0) for the control group compared to the LT group.144 The use of time as a selection tool has also been reported. UNOS guidelines require patients with liver metastasis to be free of other sites of progression by 6 months before listing for LT. However, some agree that patients with indolent progression probably do not achieve the greatest survival benefit from LT.145 It is also still not clear whether patients with more aggressive disease would benefit from LT given their lower probability of benefit from other therapies. Those are questions that need to be addressed by future research in the field.
Hepatic epithelioid haemangioendothelioma
Due to its rarity, the management of hepatic epithelioid haemangioendothelioma (HEHE) is still not well established. Furthermore, HEHE natural history varies from indolent to rapidly progressive disease.146 For instance, the 5-year OS rate is reported to be 50% after LR and 30% with systemic chemotherapy.147 In 2006, Mehrabi et al. have reviewed the published series of HEHE to date.148 They identified 434 patients with HEHE, of those nearly 45% underwent LT with a 5-year OS rate of 54.5%.148 The 5-year OS rates of patients with HEHE who underwent watchful waiting, systemic chemotherapy or radiation therapy and LR were 4.5%, 30% and 75%, respectively. In 2018, Konstantinidis et al. compared 91 patients with HEHE who underwent LR to 40 LT patients. Not surprisingly, patients in the LT group had more advanced disease (tumour size 44.6 cm in LT vs. 14.8 cm in resection) and positive lymph nodes (76.5% in LT vs. 15.4% in resection). Despite the more advanced disease, patients who underwent LT had better (but not statistically significant) OS when compared to patients treated by resection (median OS 97 months after LT and 90.5 after resection, p = 0.06).149 Lerut et al. have published the ELITA series of 59 patients with HEHE who underwent LT, 96% with bilobar disease.150 In this series, the 5- and 10-year OS rates were 83% and 72%, respectively. More recently, this European experience was expanded by Lai et al..147 The 5- and 10-year OS rates were 77% and 74%, respectively. The risk factors for recurrence were presence of macrovascular invasion, waiting time greater than 120 days and presence of lymph nodal invasion.147 A polish group reported a 3-year OS rate of 87% after LT.151 A study from the UNOS database on 110 transplanted patients with HEHE showed 5-year OS and DFS rates of 70% and 55%, respectively.152 The best management of HEHE is still to be defined, but LT might offer a survival benefit for these patients compared to other therapies. Controversies around the best criteria for patient selection and pre- and post-LT management need to be addressed by further investigations.
Conclusion
In conclusion, the field of LT is evolving rapidly to expand the indications of LT for patients with primary and secondary liver cancer. Fig. 1 presents a summary of the most important advances in LT for cancer. The current results are promising; however, caution should be taken when expanding LT criteria for cancer patients, to avoid compromising patients awaiting LT for chronic liver diseases. In the context of improvements in preoperative selection criteria, surgical technique and post-LT care, the dismal results from previous decades are not currently valid. Moreover, the better treatment of patients with chronic liver diseases (e.g. DAAs for HCV infection) will reduce the number of patients on the waiting list because of end-stage liver disease. Furthermore, techniques for donor pool expansion (e.g. donation after cardiac death, live donation, etc.) will likely improve the imbalance between the number of available grafts and the number of patients on the waiting list. In the era of transplant oncology, surgeons, hepatologists, radiation and medical oncologists should work towards the careful expansion of the use of LT for cancer patients.
Fig. 1.
Timeline of greatest advances in transplant for cancer.
Abbreviations
AFP, alpha-fetoprotein; AFP-L3, alpha-fetoprotein-lecithin 3; BMI, body mass index; CCA, cholangiocarcinoma; CEA, carcinoembryonic antigen; CRC, colorectal cancer; CRLM, colorectal liver metastasis; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; DAA, direct-acting antiviral; DCP, des-gamma-carboxy prothrombin; DFS, disease-free-survival; GALAD, acronym for: Gender, Age, AFP-L3, AFP, DCP; HCC, hepatocellular carcinoma; LRT, locoregional therapy; LT, liver transplant; OS, overall survival; PD-1, programmed cell death protein 1; PDL-1, programmed death-ligand 1; SIRT, selective internal radiation therapy; TACE, transarterial chemoembolisation; TIM-3, T-cell immunoglobulin and mucin-domain containing-3; TTD, total tumour diameter; TTV, total tumour volume; UCSF, University of California at San Francisco.
Financial support
The authors received no financial support to produce this manuscript.
Conflict of interest
The authors declare no conflicts of interest that pertain to this work.
Please refer to the accompanying ICMJE disclosure forms for further details.
Footnotes
Supplementary data to this article can be found online at https://doi.org/10.1016/j.jhepr.2019.07.004.
Supplementary data
Supplementary material
References
- 1.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 2.Engstrand J, Nilsson H, Strömberg C, Jonas E, Freedman J. Colorectal cancer liver metastases - a population-based study on incidence, management and survival. BMC Cancer. 2018;18:78. doi: 10.1186/s12885-017-3925-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Starzl TE, Putnam CW. Surgical approaches to primary and metastatic liver neoplasms. Int J Radiat Oncol Biol Phys. 1976;1:959–964. doi: 10.1016/0360-3016(76)90123-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Koneru B, Cassavilla A, Bowman J, Iwatsuki S, Starzl TE. Liver transplantation for malignant tumors. Gastroenterol Clin N Am. 1988;17:177–193. [PMC free article] [PubMed] [Google Scholar]
- 5.Penn I. Hepatic transplantation for primary and metastatic cancers of the liver. Surgery. 1991;110:726–734. discussion 34-5. [PubMed] [Google Scholar]
- 6.Iwatsuki S, Gordon RD, Shaw BW, Starzl TE. Role of liver transplantation in cancer therapy. Ann Surg. 1985;202:401–407. doi: 10.1097/00000658-198510000-00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med. 1996;334:693–699. doi: 10.1056/NEJM199603143341104. [DOI] [PubMed] [Google Scholar]
- 8.Hibi T, Sapisochin G. What is transplant oncology? Surgery. 2019;165:281–285. doi: 10.1016/j.surg.2018.10.024. [DOI] [PubMed] [Google Scholar]
- 9.Pinna AD, Yang T, Mazzaferro V, De Carlis L, Zhou J, Roayaie S. Liver Transplantation and Hepatic Resection can Achieve Cure for Hepatocellular Carcinoma. Ann Surg. 2018;268:868–875. doi: 10.1097/SLA.0000000000002889. [DOI] [PubMed] [Google Scholar]
- 10.Llovet JM. Expanding HCC criteria for liver transplant: the urgent need for prospective, robust data. Liver Transpl. 2006;12:1741–1743. doi: 10.1002/lt.20908. [DOI] [PubMed] [Google Scholar]
- 11.Sapisochin G, Bruix J. Liver transplantation for hepatocellular carcinoma: outcomes and novel surgical approaches. Nat Rev Gastroenterol Hepatol. 2017;14:203–217. doi: 10.1038/nrgastro.2016.193. [DOI] [PubMed] [Google Scholar]
- 12.Hameed B, Mehta N, Sapisochin G, Roberts JP, Yao FY. Alpha-fetoprotein level ≫ 1000 ng/ml as an exclusion criterion for liver transplantation in patients with hepatocellular carcinoma meeting the Milan criteria. Liver Transpl. 2014;20:945–951. doi: 10.1002/lt.23904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Toso C, Trotter J, Wei A, Bigam DL, Shah S, Lancaster J. Total tumor volume predicts risk of recurrence following liver transplantation in patients with hepatocellular carcinoma. Liver Transpl. 2008;14:1107–1115. doi: 10.1002/lt.21484. [DOI] [PubMed] [Google Scholar]
- 14.Toso C, Dupuis-Lozeron E, Majno P, Berney T, Kneteman NM, Perneger T. A model for dropout assessment of candidates with or without hepatocellular carcinoma on a common liver transplant waiting list. Hepatology. 2012;56:149–156. doi: 10.1002/hep.25603. [DOI] [PubMed] [Google Scholar]
- 15.Duvoux C, Roudot-Thoraval F, Decaens T, Pessione F, Badran H, Piardi T. Liver transplantation for hepatocellular carcinoma: a model including α-fetoprotein improves the performance of Milan criteria. Gastroenterology. 2012;143 doi: 10.1053/j.gastro.2012.05.052. 986-94.e3; quiz e14-5. [DOI] [PubMed] [Google Scholar]
- 16.Mazzaferro V, Sposito C, Zhou J, Pinna AD, De Carlis L, Fan J. Metroticket 2.0 Model for Analysis of Competing Risks of Death After Liver Transplantation for Hepatocellular Carcinoma. Gastroenterology. 2018;154:128–139. doi: 10.1053/j.gastro.2017.09.025. [DOI] [PubMed] [Google Scholar]
- 17.Halazun KJ, Tabrizian P, Najjar M, Florman S, Schwartz M, Michelassi F. Is it Time to Abandon the Milan Criteria?: Results of a Bicoastal US Collaboration to Redefine Hepatocellular Carcinoma Liver Transplantation Selection Policies. Ann Surg. 2018;268:690–699. doi: 10.1097/SLA.0000000000002964. [DOI] [PubMed] [Google Scholar]
- 18.DuBay D, Sandroussi C, Sandhu L, Cleary S, Guba M, Cattral MS. Liver transplantation for advanced hepatocellular carcinoma using poor tumor differentiation on biopsy as an exclusion criterion. Ann Surg. 2011;253:166–172. doi: 10.1097/sla.0b013e31820508f1. [DOI] [PubMed] [Google Scholar]
- 19.Cillo U, Vitale A, Bassanello M, Boccagni P, Brolese A, Zanus G. Liver transplantation for the treatment of moderately or well-differentiated hepatocellular carcinoma. Ann Surg. 2004;239:150–159. doi: 10.1097/01.sla.0000109146.72827.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sapisochin G, Goldaracena N, Laurence JM, Dib M, Barbas A, Ghanekar A. The extended Toronto criteria for liver transplantation in patients with hepatocellular carcinoma: A prospective validation study. Hepatology. 2016;64:2077–2088. doi: 10.1002/hep.28643. [DOI] [PubMed] [Google Scholar]
- 21.Kaido T, Ogawa K, Mori A, Fujimoto Y, Ito T, Tomiyama K. Usefulness of the Kyoto criteria as expanded selection criteria for liver transplantation for hepatocellular carcinoma. Surgery. 2013;154:1053–1060. doi: 10.1016/j.surg.2013.04.056. [DOI] [PubMed] [Google Scholar]
- 22.Shimamura T, Akamatsu N, Fujiyoshi M, Kawaguchi A, Morita S, Kawasaki S. Expanded living-donor liver transplantation criteria for patients with hepatocellular carcinoma based on the Japanese nationwide survey: the 5-5-500 rule - a retrospective study. Transpl Int. 2019;32:356–368. doi: 10.1111/tri.13391. [DOI] [PubMed] [Google Scholar]
- 23.Yang SH, Suh KS, Lee HW, Cho EH, Cho JY, Cho YB. The role of (18)F-FDG-PET imaging for the selection of liver transplantation candidates among hepatocellular carcinoma patients. Liver Transpl. 2006;12:1655–1660. doi: 10.1002/lt.20861. [DOI] [PubMed] [Google Scholar]
- 24.Hong G, Suh KS, Suh SW, Yoo T, Kim H, Park MS. Alpha-fetoprotein and (18)F-FDG positron emission tomography predict tumor recurrence better than Milan criteria in living donor liver transplantation. J Hepatol. 2016;64:852–859. doi: 10.1016/j.jhep.2015.11.033. [DOI] [PubMed] [Google Scholar]
- 25.Halazun KJ, Patzer RE, Rana AA, Verna EC, Griesemer AD, Parsons RF. Standing the test of time: outcomes of a decade of prioritizing patients with hepatocellular carcinoma, results of the UNOS natural geographic experiment. Hepatology. 2014;60:1957–1962. doi: 10.1002/hep.27272. [DOI] [PubMed] [Google Scholar]
- 26.OPTN/UNOS Organ Procurement Transplant Network and United Organ Sharing Network Policy - Liver and Intestinal Organ Transplantation Committee 2019. https://optn.transplant.hrsa.gov/media/2816/liver_nlrb-revised-policy-notice-dsa_01252019.pdf Available from:
- 27.Firl DJ, Sasaki K, Agopian VG, Gorgen A, Kimura S, Dumronggittigule W. Charting the Path Forward for Risk Prediction in Liver Transplant for Hepatocellular Carcinoma: International Validation of HALTHCC Among 4,089 Patients. Hepatology. 2019 doi: 10.1002/hep.30838. [DOI] [PubMed] [Google Scholar]
- 28.Agopian VG, Harlander-Locke MP, Ruiz RM, Klintmalm GB, Senguttuvan S, Florman SS. Impact of Pretransplant Bridging Locoregional Therapy for Patients With Hepatocellular Carcinoma Within Milan Criteria Undergoing Liver Transplantation: Analysis of 3601 Patients From the US Multicenter HCC Transplant Consortium. Ann Surg. 2017;266:525–535. doi: 10.1097/SLA.0000000000002381. [DOI] [PubMed] [Google Scholar]
- 29.Gordic S, Corcuera-Solano I, Stueck A, Besa C, Argiriadi P, Guniganti P. Evaluation of HCC response to locoregional therapy: Validation of MRI-based response criteria versus explant pathology. J Hepatol. 2017;67:1213–1221. doi: 10.1016/j.jhep.2017.07.030. [DOI] [PubMed] [Google Scholar]
- 30.DiNorcia J, Florman SS, Haydel B, Tabrizian P, Ruiz RM, Klintmalm GB. Pathologic Response to Pretransplant Locoregional Therapy is Predictive of Patient Outcome After Liver Transplantation for Hepatocellular Carcinoma: Analysis From the US Multicenter HCC Transplant Consortium. Ann Surg. 2019 doi: 10.1097/SLA.0000000000003253. [DOI] [PubMed] [Google Scholar]
- 31.Lai Q, Vitale A, Iesari S, Finkenstedt A, Mennini G, Onali S. The Intention-to-Treat Effect of Bridging Treatments in the Setting of Milan Criteria-In Patients Waiting for Liver Transplantation. Liver Transpl. 2019;25:1023–1033. doi: 10.1002/lt.25492. [DOI] [PubMed] [Google Scholar]
- 32.Mehta N, Dodge JL, Roberts JP, Hirose R, Yao FY. Alpha-Fetoprotein Decrease from ≫ 1,000 to ≪ 500 ng/ml in Patients with Hepatocellular Carcinoma Leads to Improved Posttransplant Outcomes. Hepatology. 2019;69:1193–1205. doi: 10.1002/hep.30413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhou Y, He L, Huang Y, Chen S, Wu P, Ye W. CT-based radiomics signature: a potential biomarker for preoperative prediction of early recurrence in hepatocellular carcinoma. Abdom Radiol. 2017;42:1695–1704. doi: 10.1007/s00261-017-1072-0. [DOI] [PubMed] [Google Scholar]
- 34.Kim J, Choi SJ, Lee SH, Lee HY, Park H. Predicting Survival Using Pretreatment CT for Patients With Hepatocellular Carcinoma Treated With Transarterial Chemoembolization: Comparison of Models Using Radiomics. AJR Am J Roentgenol. 2018;211:1026–1034. doi: 10.2214/AJR.18.19507. [DOI] [PubMed] [Google Scholar]
- 35.Xu X, Zhang HL, Liu QP, Sun SW, Zhang J, Zhu FP. Radiomic analysis of contrast-enhanced CT predicts microvascular invasion and outcome in hepatocellular carcinoma. J Hepatol. 2019;70(6):1133–1144. doi: 10.1016/j.jhep.2019.02.023. [DOI] [PubMed] [Google Scholar]
- 36.Bhangui P, Allard MA, Vibert E, Cherqui D, Pelletier G, Cunha AS. Salvage Versus Primary Liver Transplantation for Early Hepatocellular Carcinoma: Do Both Strategies Yield Similar Outcomes? Ann Surg. 2016;264:155–163. doi: 10.1097/SLA.0000000000001442. [DOI] [PubMed] [Google Scholar]
- 37.Muaddi H, Al-Adra DP, Beecroft R, Ghanekar A, Moulton CA, Doyle A. Liver Transplantation is Equally Effective as a Salvage Therapy for Patients with Hepatocellular Carcinoma Recurrence Following Radiofrequency Ablation or Liver Resection with Curative Intent. Ann Surg Oncol. 2018;25:991–999. doi: 10.1245/s10434-017-6329-x. [DOI] [PubMed] [Google Scholar]
- 38.Ma KW, Chok KSH, She WH, Chan ACY, Cheung TT, Dai WC. Defining Optimal Surgical Treatment for Recurrent Hepatocellular Carcinoma: A Propensity Score Matched Analysis. Liver Transpl. 2018;24:1062–1069. doi: 10.1002/lt.25033. [DOI] [PubMed] [Google Scholar]
- 39.Lim C, Shinkawa H, Hasegawa K, Bhangui P, Salloum C, Gomez Gavara C. Salvage liver transplantation or repeat hepatectomy for recurrent hepatocellular carcinoma: An intent-to-treat analysis. Liver Transpl. 2017;23:1553–1563. doi: 10.1002/lt.24952. [DOI] [PubMed] [Google Scholar]
- 40.Mehta N, Dodge JL, Roberts JP, Hirose R, Yao FY. Alpha-fetoprotein Decrease from ≫1000 to ≪500 ng/ml in Patients with Hepatocellular Carcinoma Leads to Improved Post-Transplant Outcomes. Hepatology. 2018;415 doi: 10.1002/hep.30413. hep.30413-hep. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.MacParland SA, Liu JC, Ma X-Z, Innes BT, Bartczak AM, Gage BK. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat Commun. 2018;9:4383. doi: 10.1038/s41467-018-06318-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Fedarko NS, Jain A, Karadag A, Van Eman MR, Fisher LW. Elevated serum bone sialoprotein and osteopontin in colon, breast, prostate, and lung cancer. Clin Cancer Res. 2001;7:4060–4066. [PubMed] [Google Scholar]
- 43.Berhane S, Toyoda H, Tada T, Kumada T, Kagebayashi C, Satomura S. Role of the GALAD and BALAD-2 Serologic Models in Diagnosis of Hepatocellular Carcinoma and Prediction of Survival in Patients. Clin Gastroenterol Hepatol. 2016;14 doi: 10.1016/j.cgh.2015.12.042. 875-86.e6. [DOI] [PubMed] [Google Scholar]
- 44.Vongsuvanh R, van der Poorten D, Iseli T, Strasser SI, McCaughan GW, George J. Midkine Increases Diagnostic Yield in AFP Negative and NASH-Related Hepatocellular Carcinoma. PloS one. 2016;11 doi: 10.1371/journal.pone.0155800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Marrero JA, Feng Z, Wang Y, Nguyen MH, Befeler AS, Roberts LR. α-Fetoprotein, Des-γ Carboxyprothrombin, and Lectin-Bound α-Fetoprotein in Early Hepatocellular Carcinoma. Gastroenterology. 2009;137:110–118. doi: 10.1053/j.gastro.2009.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Qiao S-S, Cui Z-Q-Q, Gong L, Han H, Chen P-C, Guo L-M. Simultaneous measurements of serum AFP, GPC-3 and HCCR for diagnosing hepatocellular carcinoma. Hepato-Gastroenterology. 2011;58:1718–1724. doi: 10.5754/hge11124. [DOI] [PubMed] [Google Scholar]
- 47.Giardina MG, Matarazzo M, Varriale A, Morante R, Napoli A, Martino R. Serum alpha-L-fucosidase. A useful marker in the diagnosis of hepatocellular carcinoma. Cancer. 1992;70:1044–1048. doi: 10.1002/1097-0142(19920901)70:5<1044::aid-cncr2820700506>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
- 48.Ismail MM, Morsi HK, Abdulateef NAB, Noaman MK, Abou El-Ella GA. Evaluation of prothrombin induced by vitamin K absence, macrophage migration inhibitory factor and Golgi protein-73 versus alpha fetoprotein for hepatocellular carcinoma diagnosis and surveillance. Scand J Clin Lab Invest. 2017;77:175–183. doi: 10.1080/00365513.2017.1286684. [DOI] [PubMed] [Google Scholar]
- 49.Pozzan C, Cardin R, Piciocchi M, Cazzagon N, Maddalo G, Vanin V. Diagnostic and prognostic role of SCCA-IgM serum levels in hepatocellular carcinoma (HCC) J Gastroenterol Hepatol. 2014;29:1637–1644. doi: 10.1111/jgh.12576. [DOI] [PubMed] [Google Scholar]
- 50.Shi K-Q, Lin Z, Chen X-J, Song M, Wang Y-Q, Cai Y-J. Hepatocellular carcinoma associated microRNA expression signature: integrated bioinformatics analysis, experimental validation and clinical significance. Oncotarget. 2015;6:25093–25108. doi: 10.18632/oncotarget.4437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wang M, Sanda M, Comunale MA, Herrera H, Swindell C, Kono Y. Changes in the Glycosylation of Kininogen and the Development of a Kininogen-Based Algorithm for the Early Detection of HCC. Cancer Epidemiol Biomarkers Prev. 2017;26:795–803. doi: 10.1158/1055-9965.EPI-16-0974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Wang X, Zhang A, Sun H. Power of metabolomics in diagnosis and biomarker discovery of hepatocellular carcinoma. Hepatology (Baltimore, Md) 2013;57:2072–2077. doi: 10.1002/hep.26130. [DOI] [PubMed] [Google Scholar]
- 53.Sengupta S, Parikh ND. Biomarker development for hepatocellular carcinoma early detection: current and future perspectives. Hepatic Oncol. 2017;4:111–122. doi: 10.2217/hep-2017-0019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Pantel K, Alix-Panabières C. Circulating tumour cells and cell-free DNA in gastrointestinal cancer. Nat Rev Gastroenterol Hepatol. 2017;14:73–74. doi: 10.1038/nrgastro.2016.198. [DOI] [PubMed] [Google Scholar]
- 55.Pellegrino R, Calvisi DF, Ladu S, Ehemann V, Staniscia T, Evert M. Oncogenic and tumor suppressive roles of polo-like kinases in human hepatocellular carcinoma. Hepatology. 2010;51:857–868. doi: 10.1002/hep.23467. [DOI] [PubMed] [Google Scholar]
- 56.Li Z, Li N, Li F, Zhou Z, Sang J, Chen Y. Immune checkpoint proteins PD-1 and TIM-3 are both highly expressed in liver tissues and correlate with their gene polymorphisms in patients with HBV-related hepatocellular carcinoma. Medicine. 2016;95 doi: 10.1097/MD.0000000000005749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sotiropoulos GC, Paul A, Molmenti E, Lang H, Frilling A, Napieralski BP. Liver transplantation for hepatocellular carcinoma in cirrhosis within the Eurotransplant area: an additional option with "livers that nobody wants". Transplantation. 2005;80:897–902. doi: 10.1097/01.tp.0000173644.63692.dc. [DOI] [PubMed] [Google Scholar]
- 58.Croome KP, Wall W, Chandok N, Beck G, Marotta P, Hernandez-Alejandro R. Inferior survival in liver transplant recipients with hepatocellular carcinoma receiving donation after cardiac death liver allografts. Liver Transpl. 2013;19:1214–1223. doi: 10.1002/lt.23715. [DOI] [PubMed] [Google Scholar]
- 59.van der Bilt JD, Kranenburg O, Nijkamp MW, Smakman N, Veenendaal LM, Te Velde EA. Ischemia/reperfusion accelerates the outgrowth of hepatic micrometastases in a highly standardized murine model. Hepatology. 2005;42:165–175. doi: 10.1002/hep.20739. [DOI] [PubMed] [Google Scholar]
- 60.Croome KP, Lee DD, Burns JM, Musto K, Paz D, Nguyen JH. The Use of Donation After Cardiac Death Allografts Does Not Increase Recurrence of Hepatocellular Carcinoma. Am J Transplant. 2015;15:2704–2711. doi: 10.1111/ajt.13306. [DOI] [PubMed] [Google Scholar]
- 61.Khorsandi SE, Yip VS, Cortes M, Jassem W, Quaglia A, O'Grady J. Does Donation After Cardiac Death Utilization Adversely Affect Hepatocellular Cancer Survival? Transplantation. 2016;100:1916–1924. doi: 10.1097/TP.0000000000001150. [DOI] [PubMed] [Google Scholar]
- 62.Kollmann D, Sapisochin G, Goldaracena N, Hansen BE, Rajakumar R, Selzner N. Expanding the donor pool: Donation after circulatory death and living liver donation do not compromise the results of liver transplantation. Liver Transpl. 2018;24:779–789. doi: 10.1002/lt.25068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Orci LA, Berney T, Majno PE, Lacotte S, Oldani G, Morel P. Donor characteristics and risk of hepatocellular carcinoma recurrence after liver transplantation. Br J Surg. 2015;102:1250–1257. doi: 10.1002/bjs.9868. [DOI] [PubMed] [Google Scholar]
- 64.Lué A, Solanas E, Baptista P, Lorente S, Araiz JJ, Garcia-Gil A. How important is donor age in liver transplantation? World J Gastroenterol. 2016;22:4966–4976. doi: 10.3748/wjg.v22.i21.4966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Haugen CE, Bowring MG, Holscher CM, Jackson KR, Garonzik-Wang J, Cameron AM. Survival benefit of accepting livers from deceased donors over 70 years old. Am J Transplant. 2019;19(7):2020–2028. doi: 10.1111/ajt.15250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Marroquin CE, Marino G, Kuo PC, Plotkin JS, Rustgi VK, Lu AD. Transplantation of hepatitis C-positive livers in hepatitis C-positive patients is equivalent to transplanting hepatitis C-negative livers. Liver Transpl. 2001;7:762–768. doi: 10.1053/jlts.2001.27088. [DOI] [PubMed] [Google Scholar]
- 67.Saab S, Ghobrial RM, Ibrahim AB, Kunder G, Durazo F, Han S. Hepatitis C positive grafts may be used in orthotopic liver transplantation: a matched analysis. Am J Transplant. 2003;3:1167–1172. doi: 10.1034/j.1600-6143.2003.00189.x. [DOI] [PubMed] [Google Scholar]
- 68.Cotter TG, Paul S, Sandıkçı B, Couri T, Bodzin AS, Little EC. Increasing Utilization and Excellent Initial Outcomes Following Liver Transplant of Hepatitis C Virus (HCV)-Viremic Donors Into HCV-Negative Recipients: Outcomes Following Liver Transplant of HCV-Viremic Donors. Hepatology. 2019;69:2381–2395. doi: 10.1002/hep.30540. [DOI] [PubMed] [Google Scholar]
- 69.Ji F, Yeo YH, Wei MT, Ogawa E, Enomoto M, Lee DH. Sustained virologic response to direct-acting antiviral therapy in patients with chronic hepatitis C and hepatocellular carcinoma: A systematic review and meta-analysis. J Hepatol. 2019;71(3):473–485. doi: 10.1016/j.jhep.2019.04.017. [DOI] [PubMed] [Google Scholar]
- 70.Carrat F, Fontaine H, Dorival C, Simony M, Diallo A, Hezode C. Clinical outcomes in patients with chronic hepatitis C after direct-acting antiviral treatment: a prospective cohort study. Lancet. 2019;393:1453–1464. doi: 10.1016/S0140-6736(18)32111-1. [DOI] [PubMed] [Google Scholar]
- 71.Cotter TG, Paul S, Sandıkçı B, Couri T, Bodzin AS, Little EC. Improved Graft Survival After Liver Transplantation for Recipients With Hepatitis C Virus in the Direct-Acting Antiviral Era. Liver Transpl. 2019;25:598–609. doi: 10.1002/lt.25424. [DOI] [PubMed] [Google Scholar]
- 72.Goldberg D, Ditah IC, Saeian K, Lalehzari M, Aronsohn A, Gorospe EC. Changes in the Prevalence of Hepatitis C Virus Infection, Nonalcoholic Steatohepatitis, and Alcoholic Liver Disease Among Patients With Cirrhosis or Liver Failure on the Waitlist for Liver Transplantation. Gastroenterology. 2017;152 doi: 10.1053/j.gastro.2017.01.003. 1090-9.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kim D, Li AA, Perumpail BJ, Gadiparthi C, Kim W, Cholankeril G. Changing Trends in Etiology-Based and Ethnicity-Based Annual Mortality Rates of Cirrhosis and Hepatocellular Carcinoma in the United States. Hepatology. 2019;69:1064–1074. doi: 10.1002/hep.30161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Belli LS, Perricone G, Adam R, Cortesi PA, Strazzabosco M, Facchetti R. Impact of DAAs on liver transplantation: Major effects on the evolution of indications and results. An ELITA study based on the ELTR registry. J Hepatol. 2018;69(4):810–817. doi: 10.1016/j.jhep.2018.06.010. [DOI] [PubMed] [Google Scholar]
- 75.Kulik LM, Fisher RA, Rodrigo DR, Brown RS, Freise CE, Shaked A. Outcomes of living and deceased donor liver transplant recipients with hepatocellular carcinoma: results of the A2ALL cohort. Am J Transplant. 2012;12:2997–3007. doi: 10.1111/j.1600-6143.2012.04272.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Fisher RA, Kulik LM, Freise CE, Lok AS, Shearon TH, Brown RS. Hepatocellular carcinoma recurrence and death following living and deceased donor liver transplantation. Am J Transplant. 2007;7:1601–1608. doi: 10.1111/j.1600-6143.2007.01802.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bhangui P, Vibert E, Majno P, Salloum C, Andreani P, Zocrato J. Intention-to-treat analysis of liver transplantation for hepatocellular carcinoma: living versus deceased donor transplantation. Hepatology. 2011;53:1570–1579. doi: 10.1002/hep.24231. [DOI] [PubMed] [Google Scholar]
- 78.Azoulay D, Audureau E, Bhangui P, Belghiti J, Boillot O, Andreani P. Living or Brain-dead Donor Liver Transplantation for Hepatocellular Carcinoma: A Multicenter, Western, Intent-to-treat Cohort Study. Ann Surg. 2017;266:1035–1044. doi: 10.1097/SLA.0000000000001986. [DOI] [PubMed] [Google Scholar]
- 79.Goldaracena N, Gorgen A, Doyle A, Hansen BE, Tomiyama K, Zhang W. Live donor liver transplantation for patients with hepatocellular carcinoma offers increased survival vs. deceased donation. J Hepatol. 2019;70:666–673. doi: 10.1016/j.jhep.2018.12.029. [DOI] [PubMed] [Google Scholar]
- 80.Rössler F, Sapisochin G, Song G, Lin YH, Simpson MA, Hasegawa K. Defining Benchmarks for Major Liver Surgery: A multicenter Analysis of 5202 Living Liver Donors. Ann Surg. 2016;264:492–500. doi: 10.1097/SLA.0000000000001849. [DOI] [PubMed] [Google Scholar]
- 81.Gorgen A, Goldaracena N, Zhang W, Rosales R, Ghanekar A, Lilly L. Surgical Complications after Right Hepatectomy for Live Liver Donation: Largest Single-Center Western World Experience. Semin Liver Dis. 2018;38:134–144. doi: 10.1055/s-0038-1636932. [DOI] [PubMed] [Google Scholar]
- 82.Trotter JF, Adam R, Lo CM, Kenison J. Documented deaths of hepatic lobe donors for living donor liver transplantation. Liver Transpl. 2006;12:1485–1488. doi: 10.1002/lt.20875. [DOI] [PubMed] [Google Scholar]
- 83.Cheah YL, Simpson MA, Pomposelli JJ, Pomfret EA. Incidence of death and potentially life-threatening near-miss events in living donor hepatic lobectomy: a world-wide survey. Liver Transpl. 2013;19:499–506. doi: 10.1002/lt.23575. [DOI] [PubMed] [Google Scholar]
- 84.Shen L, Xi M, Zhao L, Zhang X, Wang X, Huang Z. Combination Therapy after TACE for Hepatocellular Carcinoma with Macroscopic Vascular Invasion: Stereotactic Body Radiotherapy versus Sorafenib. Cancers (Basel) 2018;10 doi: 10.3390/cancers10120516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Adam R, Karam V, Cailliez V, JG O Grady, Mirza D, Cherqui D. 2018 Annual Report of the European Liver Transplant Registry (ELTR) - 50-year evolution of liver transplantation. Transpl Int. 2018;31:1293–1317. doi: 10.1111/tri.13358. [DOI] [PubMed] [Google Scholar]
- 86.O'Grady JG, Polson RJ, Rolles K, Calne RY, Williams R. Liver transplantation for malignant disease. Results in 93 consecutive patients. Ann Surg. 1988;207:373–379. doi: 10.1097/00000658-198804000-00002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Pichlmayr R, Weimann A, Tusch G, Schlitt HJ. Indications and Role of Liver Transplantation for Malignant Tumors. Oncologist. 1997;2:164–170. [PubMed] [Google Scholar]
- 88.De Vreede I, Steers JL, Burch PA, Rosen CB, Gunderson LL, Haddock MG. Prolonged disease-free survival after orthotopic liver transplantation plus adjuvant chemoirradiation for cholangiocarcinoma. Liver Transpl. 2000;6:309–316. doi: 10.1053/lv.2000.6143. [DOI] [PubMed] [Google Scholar]
- 89.Gores GJ, Darwish Murad S, Heimbach JK, Rosen CB. Liver transplantation for perihilar cholangiocarcinoma. Dig Dis. 2013;31:126–129. doi: 10.1159/000347207. [DOI] [PubMed] [Google Scholar]
- 90.Loveday BPT, Knox JJ, Dawson LA, Metser U, Brade A, Horgan AM. Neoadjuvant hyperfractionated chemoradiation and liver transplantation for unresectable perihilar cholangiocarcinoma in Canada. J Surg Oncol. 2018;117:213–219. doi: 10.1002/jso.24833. [DOI] [PubMed] [Google Scholar]
- 91.Darwish Murad S, Kim WR, Harnois DM, Douglas DD, Burton J, Kulik LM. Efficacy of neoadjuvant chemoradiation, followed by liver transplantation, for perihilar cholangiocarcinoma at 12 US centers. Gastroenterology. 2012;143:88–98.e3. doi: 10.1053/j.gastro.2012.04.008. quiz e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Mantel HT, Westerkamp AC, Adam R, Bennet WF, Seehofer D, Settmacher U. Strict Selection Alone of Patients Undergoing Liver Transplantation for Hilar Cholangiocarcinoma Is Associated with Improved Survival. PLoS One. 2016;11 doi: 10.1371/journal.pone.0156127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.OPTN PAolal-i Organ Procurement and Transplantation Network 12/05/2018. https://optn.transplant.hrsa.gov/media/1200/optn_policies.pdf#nameddest=Policy_09 Available from:
- 94.Trillium Ontario's Adult Referral and Listing Criteria for Liver Transplantation Toronto, ON, Canada: Trillium Gift of Life Network. 2017. https://www.giftoflife.on.ca/resources/pdf/5_Adult_Liver_TxRef_List_Criteria_Dec417.pdf Available from:
- 95.Eurotransplant Eurotransplant manual - Chapter 5: ET Liver Allocation System (ELAS): Eurotransplant. 2018. http://www.eurotransplant.org/cms/mediaobject.php?file=H5+ELAS+MELD+November+20181.pdf Available from:
- 96.Rosen CB, Darwish Murad S, Heimbach JK, Nyberg SL, Nagorney DM, Gores GJ. Neoadjuvant therapy and liver transplantation for hilar cholangiocarcinoma: is pretreatment pathological confirmation of diagnosis necessary? J Am Coll Surg. 2012;215:31–38. doi: 10.1016/j.jamcollsurg.2012.03.014. discussion 8-40. [DOI] [PubMed] [Google Scholar]
- 97.Duignan S, Maguire D, Ravichand CS, Geoghegan J, Hoti E, Fennelly D. Neoadjuvant chemoradiotherapy followed by liver transplantation for unresectable cholangiocarcinoma: a single-centre national experience. HPB (Oxford) 2014;16:91–98. doi: 10.1111/hpb.12082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hong JC, Jones CM, Duffy JP, Petrowsky H, Farmer DG, French S. Comparative analysis of resection and liver transplantation for intrahepatic and hilar cholangiocarcinoma: a 24-year experience in a single center. Arch Surg. 2011;146:683–689. doi: 10.1001/archsurg.2011.116. [DOI] [PubMed] [Google Scholar]
- 99.Ebata T, Mizuno T, Yokoyama Y, Igami T, Sugawara G, Nagino M. Surgical resection for Bismuth type IV perihilar cholangiocarcinoma. Br J Surg. 2018;105:829–838. doi: 10.1002/bjs.10556. [DOI] [PubMed] [Google Scholar]
- 100.Ethun CG, Lopez-Aguiar AG, Anderson DJ, Adams AB, Fields RC, Doyle MB. Transplantation Versus Resection for Hilar Cholangiocarcinoma: An Argument for Shifting Treatment Paradigms for Resectable Disease. Ann Surg. 2018;267:797–805. doi: 10.1097/SLA.0000000000002574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Vibert E. ClinicalTrialsgov [internet] National Library of Medicine (US); Bethesda (MD): 2000. Randomized Prospective Multicentric Study: Radio-chemotherapy and Liver Transplantation Versus Liver Resection to Treat Respectable Hilar Cholangiocarcinoma Paul Brousse Hospital, Villejuif. [ NCT02232932:[ https://clinicaltrials.gov/ct2/show/NCT02232932?term=TRANSPHIL&rank=1] [Google Scholar]
- 102.Robles R, Figueras J, Turrión VS, Margarit C, Moya A, Varo E. Spanish experience in liver transplantation for hilar and peripheral cholangiocarcinoma. Ann Surg. 2004;239:265–271. doi: 10.1097/01.sla.0000108702.45715.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Sapisochin G. Rodríguez de Lope C, Gastaca M, Ortiz de Urbina J, Suarez MA, Santoyo J, et al. "Very early" intrahepatic cholangiocarcinoma in cirrhotic patients: should liver transplantation be reconsidered in these patients? Am J Transplant. 2014;14:660–667. doi: 10.1111/ajt.12591. [DOI] [PubMed] [Google Scholar]
- 104.Vilchez V, Shah MB, Daily MF, Pena L, Tzeng CW, Davenport D. Long-term outcome of patients undergoing liver transplantation for mixed hepatocellular carcinoma and cholangiocarcinoma: an analysis of the UNOS database. HPB (Oxford) 2016;18:29–34. doi: 10.1016/j.hpb.2015.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Sapisochin G, Facciuto M, Rubbia-Brandt L, Marti J, Mehta N, Yao FY. Liver transplantation for "very early" intrahepatic cholangiocarcinoma: International retrospective study supporting a prospective assessment. Hepatology (Baltimore, Md) 2016;64:1178–1188. doi: 10.1002/hep.28744. [DOI] [PubMed] [Google Scholar]
- 106.G S. University Health Network, Toronto. Liver Transplantation for Early Intrahepatic Cholangiocarcinoma University Health Network, Toronto. In: ClinicalTiralsgov [internet] Bethesda (MD): National Library of Medicine (US) 20002016 [Clinicaltrials.gov [internet] Bethesda (MD): National Library of Medicine (US) Identifier: NCT02878473]. Available from: https://clinicaltrials.gov/ct2/show/NCT02878473?term=sapisochin&rank=3.
- 107.Lunsford KE, Javle M, Heyne K, Shroff RT, Abdel-Wahab R, Gupta N. Liver transplantation for locally advanced intrahepatic cholangiocarcinoma treated with neoadjuvant therapy: a prospective case-series. Lancet Gastroenterol Hepatol. 2018;3:337–348. doi: 10.1016/S2468-1253(18)30045-1. [DOI] [PubMed] [Google Scholar]
- 108.Le Roy B, Gelli M, Pittau G, Allard MA, Pereira B, Serji B. Neoadjuvant chemotherapy for initially unresectable intrahepatic cholangiocarcinoma. Br J Surg. 2018;105:839–847. doi: 10.1002/bjs.10641. [DOI] [PubMed] [Google Scholar]
- 109.Rayar M, Sulpice L, Edeline J, Garin E, Levi Sandri GB, Meunier B. Intra-arterial yttrium-90 radioembolization combined with systemic chemotherapy is a promising method for downstaging unresectable huge intrahepatic cholangiocarcinoma to surgical treatment. Ann Surg Oncol. 2015;22:3102–3108. doi: 10.1245/s10434-014-4365-3. [DOI] [PubMed] [Google Scholar]
- 110.Baumgartner JM, Raymond VM, Lanman RB, Tran L, Kelly KJ, Lowy AM. Preoperative Circulating Tumor DNA in Patients with Peritoneal Carcinomatosis is an Independent Predictor of Progression-Free Survival. Ann Surg Oncol. 2018;25:2400–2408. doi: 10.1245/s10434-018-6561-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Simbolo M, Fassan M, Ruzzenente A, Mafficini A, Wood LD, Corbo V. Multigene mutational profiling of cholangiocarcinomas identifies actionable molecular subgroups. Oncotarget. 2014;5:2839–2852. doi: 10.18632/oncotarget.1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Ruzzenente A, Fassan M, Conci S, Simbolo M, Lawlor RT, Pedrazzani C. Cholangiocarcinoma Heterogeneity Revealed by Multigene Mutational Profiling: Clinical and Prognostic Relevance in Surgically Resected Patients. Ann Surg Oncol. 2016;23:1699–1707. doi: 10.1245/s10434-015-5046-6. [DOI] [PubMed] [Google Scholar]
- 113.Zhu AX, Borger DR, Kim Y, Cosgrove D, Ejaz A, Alexandrescu S. Genomic profiling of intrahepatic cholangiocarcinoma: refining prognosis and identifying therapeutic targets. Ann Surg Oncol. 2014;21:3827–3834. doi: 10.1245/s10434-014-3828-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Fabris L, Perugorria MJ, Mertens J, Björkström NK, Cramer T, Lleo A. The tumour microenvironment and immune milieu of cholangiocarcinoma. Liver Int. 2019;39:63–78. doi: 10.1111/liv.14098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Siegel RL, Miller KD, Jemal A. Colorectal Cancer Mortality Rates in Adults Aged 20 to 54 Years in the United States, 1970-2014. JAMA. 2017;318:572–574. doi: 10.1001/jama.2017.7630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Manfredi S, Lepage C, Hatem C, Coatmeur O, Faivre J, Bouvier AM. Epidemiology and management of liver metastases from colorectal cancer. Ann Surg. 2006;244:254–259. doi: 10.1097/01.sla.0000217629.94941.cf. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mühlbacher F, Huk I, Steininger R, Gnant M, Götzinger P, Wamser P. Is orthotopic liver transplantation a feasible treatment for secondary cancer of the liver? Transplant Proc. 1991;23:1567–1568. [PubMed] [Google Scholar]
- 118.Kappel S, Kandioler D, Steininger R, Längle F, Wrba F, Ploder M. Genetic detection of lymph node micrometastases: a selection criterion for liver transplantation in patients with liver metastases after colorectal cancer. Transplantation. 2006;81:64–70. doi: 10.1097/01.tp.0000189711.98971.9c. [DOI] [PubMed] [Google Scholar]
- 119.Hagness M, Foss A, Line PD, Scholz T, Jørgensen PF, Fosby B. Liver transplantation for nonresectable liver metastases from colorectal cancer. Ann Surg. 2013;257:800–806. doi: 10.1097/SLA.0b013e3182823957. [DOI] [PubMed] [Google Scholar]
- 120.Fosby B, Melum E, Bjøro K, Bennet W, Rasmussen A, Andersen IM. Liver transplantation in the Nordic countries - An intention to treat and post-transplant analysis from The Nordic Liver Transplant Registry 1982-2013. Scand J Gastroenterol. 2015;50:797–808. doi: 10.3109/00365521.2015.1036359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Hagness M, Foss A, Egge TS, Dueland S. Patterns of recurrence after liver transplantation for nonresectable liver metastases from colorectal cancer. Ann Surg Oncol. 2014;21:1323–1329. doi: 10.1245/s10434-013-3449-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Toso C, Pinto Marques H, Andres A, Castro Sousa F, Adam R, Kalil A. Liver transplantation for colorectal liver metastasis: Survival without recurrence can be achieved. Liver Transpl. 2017;23:1073–1076. doi: 10.1002/lt.24791. [DOI] [PubMed] [Google Scholar]
- 123.Lai Q, Vitale A, Iesari S, Finkenstedt A, Mennini G, Spoletini G. Intention-to-treat survival benefit of liver transplantation in patients with hepatocellular cancer. Hepatology. 2017;66:1910–1919. doi: 10.1002/hep.29342. [DOI] [PubMed] [Google Scholar]
- 124.Englschalk C, Eser D, Jox RJ, Gerbes A, Frey L, Dubay DA. Benefit in liver transplantation: a survey among medical staff, patients, medical students and non-medical university staff and students. BMC Med Ethics. 2018;19:7. doi: 10.1186/s12910-018-0248-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Dueland S, Guren TK, Hagness M, Glimelius B, Line PD, Pfeiffer P. Chemotherapy or liver transplantation for nonresectable liver metastases from colorectal cancer? Ann Surg. 2015;261:956–960. doi: 10.1097/SLA.0000000000000786. [DOI] [PubMed] [Google Scholar]
- 126.Bjørnelv GMW, Dueland S, Line PD, Joranger P, Fretland Å, Edwin B. Cost-effectiveness of liver transplantation in patients with colorectal metastases confined to the liver. Br J Surg. 2019;106:132–141. doi: 10.1002/bjs.10962. [DOI] [PubMed] [Google Scholar]
- 127.Dueland S, Foss A, Solheim JM, Hagness M, Line P. 2018. Survival following liver transplantation for liver-only colorectal metastases compared with hepatocellular carcinoma; pp. 736–742. [DOI] [PubMed] [Google Scholar]
- 128.Grut H, Solberg S, Seierstad T, Revheim ME, Egge TS, Larsen SG. Growth rates of pulmonary metastases after liver transplantation for unresectable colorectal liver metastases. Br J Surg. 2018;105:295–301. doi: 10.1002/bjs.10651. [DOI] [PubMed] [Google Scholar]
- 129.Grut H, Dueland S, Line PD, Revheim ME. The prognostic value of. Eur J Nucl Med Mol Imaging. 2018;45:218–225. doi: 10.1007/s00259-017-3843-9. [DOI] [PubMed] [Google Scholar]
- 130.Dueland S, Syversveen T, Solheim JM, Solberg S, Grut H, Bjørnbeth BA. Survival Following Liver Transplantation for Patients With Nonresectable Liver-only Colorectal Metastases. Ann Surg. 2019 doi: 10.1097/SLA.0000000000003404. [DOI] [PubMed] [Google Scholar]
- 131.Adam R, Wicherts Da, De Haas R, Ciacio O, Levi F, Paule B. Patients with initially unresectable colorectal liver metastases: Is there a possibility of cure? J Clin Oncol. 2009;27:1829–1835. doi: 10.1200/JCO.2008.19.9273. [DOI] [PubMed] [Google Scholar]
- 132.Masi G, Loupakis F, Pollina L, Vasile E, Cupini S, Ricci S. Long-term outcome of initially unresectable metastatic colorectal cancer patients treated with 5-fluorouracil/leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) followed by radical surgery of metastases. Ann Surg. 2009;249:420–425. doi: 10.1097/SLA.0b013e31819a0486. [DOI] [PubMed] [Google Scholar]
- 133.Line PD, Hagness M, Berstad AE, Foss A, Dueland S. A Novel Concept for Partial Liver Transplantation in Nonresectable Colorectal Liver Metastases: The RAPID Concept. Ann Surg. 2015;262:e5–e9. doi: 10.1097/SLA.0000000000001165. [DOI] [PubMed] [Google Scholar]
- 134.Ayez N, Lalmahomed ZS, Eggermont AM, Ijzermans JN, de Jonge J, van Montfort K. Outcome of microscopic incomplete resection (R1) of colorectal liver metastases in the era of neoadjuvant chemotherapy. Ann Surg Oncol. 2012;19:1618–1627. doi: 10.1245/s10434-011-2114-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.de Haas RJ, Wicherts DA, Flores E, Azoulay D, Castaing D, Adam R. R1 resection by necessity for colorectal liver metastases: is it still a contraindication to surgery? Ann Surg. 2008;248:626–637. doi: 10.1097/SLA.0b013e31818a07f1. [DOI] [PubMed] [Google Scholar]
- 136.Adam R, Yi B, Innominato PF, Barroso E, Laurent C, Giuliante F. Resection of colorectal liver metastases after second-line chemotherapy: is it worthwhile? A LiverMetSurvey analysis of 6415 patients. Eur J Cancer. 2017;78:7–15. doi: 10.1016/j.ejca.2017.03.009. [DOI] [PubMed] [Google Scholar]
- 137.Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE. One hundred years after "carcinoid": epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–3072. doi: 10.1200/JCO.2007.15.4377. [DOI] [PubMed] [Google Scholar]
- 138.Gedaly R, Daily MF, Davenport D, McHugh PP, Koch A, Angulo P. Liver transplantation for the treatment of liver metastases from neuroendocrine tumors: an analysis of the UNOS database. Arch Surg. 2011;146:953–958. doi: 10.1001/archsurg.2011.186. [DOI] [PubMed] [Google Scholar]
- 139.Frilling A, Modlin IM, Kidd M, Russell C, Breitenstein S, Salem R. Recommendations for management of patients with neuroendocrine liver metastases. Lancet Oncol. 2014;15:e8–21. doi: 10.1016/S1470-2045(13)70362-0. [DOI] [PubMed] [Google Scholar]
- 140.Mazzaferro V, Pulvirenti A, Coppa J. Neuroendocrine tumors metastatic to the liver: how to select patients for liver transplantation? J Hepatol. 2007;47:460–466. doi: 10.1016/j.jhep.2007.07.004. [DOI] [PubMed] [Google Scholar]
- 141.Orditura M, Petrillo A, Ventriglia J, Diana A, Laterza MM, Fabozzi A. Pancreatic neuroendocrine tumors: Nosography, management and treatment. Int J Surg. 2016;28:S156–S162. doi: 10.1016/j.ijsu.2015.12.052. [DOI] [PubMed] [Google Scholar]
- 142.Le Treut YP, Grégoire E, Klempnauer J, Belghiti J, Jouve E, Lerut J. Liver transplantation for neuroendocrine tumors in Europe-results and trends in patient selection: a 213-case European liver transplant registry study. Ann Surg. 2013;257:807–815. doi: 10.1097/SLA.0b013e31828ee17c. [DOI] [PubMed] [Google Scholar]
- 143.van Vilsteren FG, Baskin-Bey ES, Nagorney DM, Sanderson SO, Kremers WK, Rosen CB. Liver transplantation for gastroenteropancreatic neuroendocrine cancers: Defining selection criteria to improve survival. Liver Transpl. 2006;12:448–456. doi: 10.1002/lt.20702. [DOI] [PubMed] [Google Scholar]
- 144.Mazzaferro V, Sposito C, Coppa J, Miceli R, Bhoori S, Bongini M. The Long-Term Benefit of Liver Transplantation for Hepatic Metastases From Neuroendocrine Tumors. Am J Transplant. 2016;16:2892–2902. doi: 10.1111/ajt.13831. [DOI] [PubMed] [Google Scholar]
- 145.Fan ST, Le Treut YP, Mazzaferro V, Burroughs AK, Olausson M, Breitenstein S. Liver transplantation for neuroendocrine tumour liver metastases. HPB (Oxford) 2015;17:23–28. doi: 10.1111/hpb.12308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Otrock ZK, Al-Kutoubi A, Kattar MM, Zaatari G, Soweid A. Spontaneous complete regression of hepatic epithelioid haemangioendothelioma. Lancet Oncol. 2006;7:439–441. doi: 10.1016/S1470-2045(06)70697-0. [DOI] [PubMed] [Google Scholar]
- 147.Lai Q, Feys E, Karam V, Adam R, Klempnauer J, Oliverius M. Hepatic Epithelioid Hemangioendothelioma and Adult Liver Transplantation: Proposal for a Prognostic Score Based on the Analysis of the ELTR-ELITA Registry. Transplantation. 2017;101:555–564. doi: 10.1097/TP.0000000000001603. [DOI] [PubMed] [Google Scholar]
- 148.Mehrabi A, Kashfi A, Fonouni H, Schemmer P, Schmied BM, Hallscheidt P. Primary malignant hepatic epithelioid hemangioendothelioma: a comprehensive review of the literature with emphasis on the surgical therapy. Cancer. 2006;107:2108–2121. doi: 10.1002/cncr.22225. [DOI] [PubMed] [Google Scholar]
- 149.Konstantinidis IT, Nota C, Jutric Z, Ituarte P, Chow W, Chu P. Primary liver sarcomas in the modern era: Resection or transplantation? J Surg Oncol. 2018;117:886–891. doi: 10.1002/jso.24979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Lerut JP, Orlando G, Adam R, Schiavo M, Klempnauer J, Mirza D. The place of liver transplantation in the treatment of hepatic epitheloid hemangioendothelioma: report of the European liver transplant registry. Ann Surg. 2007;246:949–957. doi: 10.1097/SLA.0b013e31815c2a70. discussion 57. [DOI] [PubMed] [Google Scholar]
- 151.Remiszewski P, Szczerba E, Kalinowski P, Gierej B, Dudek K, Grodzicki M. Epithelioid hemangioendothelioma of the liver as a rare indication for liver transplantation. World J Gastroenterol. 2014;20:11333–11339. doi: 10.3748/wjg.v20.i32.11333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Rodriguez JA, Becker NS, O'Mahony CA, Goss JA, Aloia TA. Long-term outcomes following liver transplantation for hepatic hemangioendothelioma: the UNOS experience from 1987 to 2005. J Gastrointest Surg. 2008;12:110–116. doi: 10.1007/s11605-007-0247-3. [DOI] [PubMed] [Google Scholar]
- 153.Yao FY, Ferrell L, Bass NM, Watson JJ, Bacchetti P, Venook A. Liver transplantation for hepatocellular carcinoma: expansion of the tumor size limits does not adversely impact survival. Hepatology. 2001;33:1394–1403. doi: 10.1053/jhep.2001.24563. [DOI] [PubMed] [Google Scholar]
- 154.Mazzaferro V, Llovet JM, Miceli R, Bhoori S, Schiavo M, Mariani L. Predicting survival after liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: a retrospective, exploratory analysis. Lancet Oncol. 2009;10:35–43. doi: 10.1016/S1470-2045(08)70284-5. [DOI] [PubMed] [Google Scholar]
- 155.Jang ES, Jeong S-H, Kim J-W, Choi YS, Leissner P, Brechot C. Diagnostic Performance of Alpha-Fetoprotein, Protein Induced by Vitamin K Absence, Osteopontin, Dickkopf-1 and Its Combinations for Hepatocellular Carcinoma. PloS one. 2016;11 doi: 10.1371/journal.pone.0151069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Sangro B, Gomez-Martin C, De La Mata M, Iñarrairaegui M, Garralda E, Barrera P. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol. 2013;59:81–88. doi: 10.1016/j.jhep.2013.02.022. [DOI] [PubMed] [Google Scholar]
- 157.Sun J, Jiang W, Tian D, Guo Q, Shen Z. Icotinib inhibits the proliferation of hepatocellular carcinoma cells in vitro and in vivo dependently on EGFR activation and PDL1 expression. OncoTargets Ther. 2018;11:8227–8237. doi: 10.2147/OTT.S179844. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplementary material

